Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
DNA Cell Biol ; 43(1): 12-25, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38170186

RESUMEN

The male sex-determining gene, sex-determining region on the Y chromosome (SRY), is expressed in adult testicular germ cells; however, its role in regulating spermatogenesis remains unclear. The role of SRY in the postmeiotic gene expression was investigated by determining the effect of SRY on the promoter of the haploid-specific Protamine 1 (PRM1) gene, which harbors five distinct SRY-binding motifs. In a luciferase reporter assay system, SRY upregulates PRM1 promoter activity in vitro in a dose-dependent manner. Through a gel-shift assay involving a 31-bp DNA fragment encompassing the SRY element within the PRM1 promoter, the third SRY-binding site on the sense strand (-373/-367) was identified as crucial for PRM1 promoter activation. This assay was extended to analyze 9 SRY variants found in the testicular DNA of 44 azoospermia patients. The findings suggest that SRY regulates PRM1 promoter activity by directly binding to its specific motif within the PRM1 promoter.


Asunto(s)
Testículo , Cromosoma Y , Humanos , Masculino , ADN/metabolismo , Protaminas/genética , Protaminas/metabolismo , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Testículo/metabolismo , Cromosoma Y/metabolismo
2.
Mol Hum Reprod ; 29(8)2023 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-37354519

RESUMEN

The Y-linked zinc finger gene ZFY is conserved across eutherians and is known to be a critical fertility factor in some species. The initial studies of the mouse homologues, Zfy1 and Zfy2, were performed using mice with spontaneous Y chromosome mutations and Zfy transgenes. These studies revealed that Zfy is involved in multiple processes during spermatogenesis, including removal of germ cells with unpaired chromosomes and control of meiotic sex chromosome inactivation during meiosis I, facilitating the progress of meiosis II, promoting spermiogenesis, and improving assisted reproduction outcomes. Zfy was also identified as a key gene in Y chromosome evolution, protecting this chromosome from extinction by serving as the executioner responsible for meiosis surveillance. Studies with targeted Zfy knock-outs revealed that mice lacking both homologues have severe spermatogenic defects and are infertile. Based on protein structure and in vitro assays, Zfy is expected to drive spermatogenesis as a transcriptional regulator. The combined evidence documents that the presence of at least one Zfy homologue is required for male fertility and that Zfy2 plays a more prominent role. This knowledge reinforces the importance of these factors for mouse spermatogenesis and informs our understanding of the human ZFY variants, which are homologous to the mouse Zfy1 and Zfy2.


Asunto(s)
Proteínas de Unión al ADN , Factores de Transcripción , Masculino , Humanos , Ratones , Animales , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/genética , Cromosoma Y/genética , Cromosoma Y/metabolismo , Espermatogénesis/genética , Dedos de Zinc/genética
3.
J Vet Med Sci ; 85(4): 507-514, 2023 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-36858585

RESUMEN

The expression of sex determining region of the Y chromosome (Sry) in the fetal gonads is important for male development. In a mouse model of disorders of sex development (C57BL/6 (B6)-XYPOS), the gonadal phenotype and the timing of Sry expression differ due to differences among B6 substrains as the genetic background. Since differences in Sry expression among B6 substrains have been speculated, the present study examined Sry expression in B6J, B6JJmsSlc, and B6NCrl mice. These substrains differed in the number of Sry-expressing cells in the gonads of embryonic mice at each developmental stage, with B6NCrl having more than the other strains. The substrains differed also in the number of Sry-expressing cells between the left and right gonads, with B6J and B6NCrl, but not B6JJmsSlc, showing left gonad-dominant Sry expression. Substrain differences existed also in the distribution of Sry-expressing cells in the medial and lateral directions of gonads. In addition, in the left gonad-dominant Sry-expressing substrains B6J and B6NCrl, the medial and central regions of the left gonad had more Sry-expressing cells than those of the right gonad. Substrains of B6 mice have not always been considered in sex differentiation studies. In the present study, however, we observed substrain differences in the number of Sry-expressing cells, left-right distribution, and medial/lateral distribution during the early stages of gonadal development in B6 mice. Therefore, future studies on sex differentiation in B6 mice should consider substrain differences.


Asunto(s)
Gónadas , Cromosoma Y , Ratones , Masculino , Animales , Ratones Endogámicos C57BL , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Cromosoma Y/genética , Cromosoma Y/metabolismo , Diferenciación Sexual/genética , Testículo/metabolismo
4.
Curr Stem Cell Res Ther ; 17(8): 750-755, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34727865

RESUMEN

Eukaryotic translation initiation factor 2 subunit 3 and structural gene Y-linked (Eif2s3y) gene, the gene encoding eIF2γ protein, is located on the mouse Y chromosome short arm. The Eif2s3y gene is globally expressed in all tissues and plays an important role in regulating global and gene-specific mRNA translation initiation. During the process of protein translation initiation, Eif2s3x (its homolog) and Eif2s3y encoded eIF2γ perform similar functions. However, it has been noticed that Eif2s3y plays a crucial role in spermatogenesis, including spermatogonia mitosis, meiosis, and spermiogenesis of spermatids, which may account for infertility. In the period of spermatogenesis, the role of Eif2s3x and Eif2s3y are not equivalent. Importance of Eif2s3y has been observed in ESC and implicated in several aspects, including the pluripotency state and the proliferation rate. Here, we discuss the functional significance of Eif2s3y in mouse spermatogenesis and self-renewal of ESCs.


Asunto(s)
Factor 2 Procariótico de Iniciación , Espermatogénesis , Animales , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Masculino , Ratones , Factor 2 Procariótico de Iniciación/genética , Espermatogénesis/genética , Espermatogonias , Factores de Transcripción/metabolismo , Cromosoma Y/metabolismo
5.
Nat Commun ; 12(1): 6854, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34824217

RESUMEN

Transposable elements (TEs) must replicate in germline cells to pass novel insertions to offspring. In Drosophila melanogaster ovaries, TEs can exploit specific developmental windows of opportunity to evade host silencing and increase their copy numbers. However, TE activity and host silencing in the distinct cell types of Drosophila testis are not well understood. Here, we reanalyze publicly available single-cell RNA-seq datasets to quantify TE expression in the distinct cell types of the Drosophila testis. We develop a method for identification of TE and host gene expression modules and find that a distinct population of early spermatocytes expresses a large number of TEs at much higher levels than other germline and somatic components of the testes. This burst of TE expression coincides with the activation of Y chromosome fertility factors and spermatocyte-specific transcriptional regulators, as well as downregulation of many components of the piRNA pathway. The TEs expressed by this cell population are specifically enriched on the Y chromosome and depleted on the X chromosome, relative to other active TEs. These data suggest that some TEs may achieve high insertional activity in males by exploiting a window of opportunity for mobilization created by the activation of spermatocyte-specific and Y chromosome-specific transcriptional programs.


Asunto(s)
Elementos Transponibles de ADN/genética , Drosophila melanogaster/genética , Espermatogénesis/genética , Cromosoma Y/genética , Animales , Drosophila melanogaster/citología , Evolución Molecular , Expresión Génica , Redes Reguladoras de Genes , Genes Ligados a Y/genética , Masculino , Mutagénesis Insercional , ARN Interferente Pequeño/genética , Espermatocitos/metabolismo , Testículo/citología , Testículo/metabolismo , Cromosoma Y/metabolismo
6.
Reprod Domest Anim ; 56(6): 928-935, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33829570

RESUMEN

The differential proteins associated with plasma membrane of spermatozoa are less known, identification of which shall help overcome limitations of currently used methods of sperm sexing, considered as a high priority for livestock sector of many countries. This study has reported plasma membrane proteomics of unsorted spermatozoa and differential expression of plasma membrane-associated proteins between X- and Y-chromosome bearing spermatozoa of indicus cattle (Bos indicus). Isolation of plasma membrane fraction using percoll gradient, relatively a rapid method, from bovine spermatozoa has been reported to enrich isolation of plasma membrane proteins. Significant enrichment for plasma membrane-associated proteins was observed in plasma membrane fraction (p < .05) as compared to the total cell lysate using LC-MS/MS. Furthermore, these experiments were conducted in flow cytometry sorted, sexed-semen samples. Thirteen proteins were identified as differentially abundant between X- and Y-sorted spermatozoa. Among these, two proteins were downregulated in Y-sorted spermatozoa compared to the X-sorted spermatozoa (p < .05), while four and seven proteins could be noted in X- and Y-sorted spermatozoa, respectively. Proteins that are presumed to support sperm capacitation and sperm migration velocity were found to be abundant in Y-sorted spermatozoa while those associated with structural molecule activity were identified as abundant in X-sorted spermatozoa in the present study. Our study provides better insight into the plasma membrane proteomics of spermatozoa of indicus cattle and furnishes data that might aid in design and development of alternate and open technology for sex-sorting of semen.


Asunto(s)
Membrana Celular/química , Proteoma/análisis , Espermatozoides/citología , Animales , Bovinos , Masculino , Preselección del Sexo/veterinaria , Espermatozoides/química , Cromosoma X/metabolismo , Cromosoma Y/metabolismo
7.
Nat Commun ; 11(1): 5537, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139741

RESUMEN

Large portions of eukaryotic genomes consist of transposable elements (TEs), and the establishment of transcription-repressing heterochromatin during early development safeguards genome integrity in Drosophila. Repeat-rich Y chromosomes can act as reservoirs for TEs ('toxic' Y effect), and incomplete epigenomic defenses during early development can lead to deleterious TE mobilization. Here, we contrast the dynamics of early TE activation in two Drosophila species with vastly different Y chromosomes of different ages. Zygotic TE expression is elevated in male embryos relative to females in both species, mostly due to expression of Y-linked TEs. Interestingly, male-biased TE expression diminishes across development in D. pseudoobscura, but remains elevated in D. miranda, the species with the younger and larger Y chromosome. The repeat-rich Y of D. miranda still contains many actively transcribed genes, which compromise the formation of silencing heterochromatin. Elevated TE expression results in more de novo insertions of repeats in males compared to females. This lends support to the idea that the 'toxic' Y chromosome can create a mutational burden in males when genome-wide defense mechanisms are compromised, and suggests a previously unappreciated epigenetic conflict on evolving Y chromosomes between transcription of essential genes and silencing of selfish DNA.


Asunto(s)
Elementos Transponibles de ADN/genética , Drosophila/genética , Silenciador del Gen , Transcripción Genética , Cromosoma Y/metabolismo , Animales , Femenino , Heterocromatina/metabolismo , Modelos Lineales , Masculino , Modelos Genéticos , Mutación , Factores Sexuales , Cigoto
8.
Nat Protoc ; 15(8): 2645-2667, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32681149

RESUMEN

The preferred sex of livestock differs among breeders; for example, dairy farmers prefer female calves for the production of milk, whereas cattle meat producers often prefer males. Sexing of laboratory animals is also beneficial in some research fields, including reproductive biology and metabolic studies. Most sexing methods separate X sperm and Y sperm with a cell sorter. Here, we describe a system in which treatment with the TLR7/8 ligand (R848) separates X sperm from Y sperm. Because this protocol does not require any special equipment or professional skills, it can be easily applied in laboratories where in vitro fertilization (IVF) is performed. The sperm are treated with 0.03 µM R848 in 1 mL of modified human tubal fluid (mHTF) medium (mouse sperm) or 3 mL of mHTF medium (bull sperm) for 60 min, and then the upper layer (400 µL in mouse sperm or 1 mL in bull sperm) and the precipitate are separately collected. After each sample is washed by centrifugation, the sperm are suspended in ligand-free IVF medium and can then be used for IVF. More than 90% of the embryos made with upper-layer sperm are XY in both mice and cattle, and >80% of the embryos made with precipitated sperm are XX in both species. Separation of X sperm and Y sperm for IVF can be completed within 2 h.


Asunto(s)
Preselección del Sexo/métodos , Espermatozoides/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Cromosoma X/metabolismo , Cromosoma Y/metabolismo , Animales , Bovinos , Femenino , Masculino , Ratones
9.
Mol Cell ; 78(3): 493-505.e8, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32353257

RESUMEN

The promyelocytic leukemia (PML) body is a phase-separated nuclear structure physically associated with chromatin, implying its crucial roles in genome functions. However, its role in transcriptional regulation is largely unknown. We developed APEX-mediated chromatin labeling and purification (ALaP) to identify the genomic regions proximal to PML bodies. We found that PML bodies associate with active regulatory regions across the genome and with ∼300 kb of the short arm of the Y chromosome (YS300) in mouse embryonic stem cells. The PML body association with YS300 is essential for the transcriptional activity of the neighboring Y-linked clustered genes. Mechanistically, PML bodies provide specific nuclear spaces that the de novo DNA methyltransferase DNMT3A cannot access, resulting in the steady maintenance of a hypo-methylated state at Y-linked gene promoters. Our study underscores a new mechanism for gene regulation in the 3D nuclear space and provides insights into the functional properties of nuclear structures for genome function.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Regulación de la Expresión Génica , Cuerpos de Inclusión Intranucleares/genética , Cromosoma Y/genética , Animales , Línea Celular , Cromatina/genética , Cromatina/metabolismo , ARN Helicasas DEAD-box/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Células Madre Embrionarias/fisiología , Endonucleasas/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Cuerpos de Inclusión Intranucleares/metabolismo , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/genética , Enzimas Multifuncionales/genética , Familia de Multigenes , Estrés Oxidativo , Proteína de la Leucemia Promielocítica/genética , Proteína de la Leucemia Promielocítica/metabolismo , Proteínas/genética , Factores de Transcripción/genética , Cromosoma Y/metabolismo
10.
BMC Genomics ; 21(1): 259, 2020 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-32228451

RESUMEN

BACKGROUND: The olive fruit fly, Bactrocera oleae, is the most important pest in the olive fruit agribusiness industry. This is because female flies lay their eggs in the unripe fruits and upon hatching the larvae feed on the fruits thus destroying them. The lack of a high-quality genome and other genomic and transcriptomic data has hindered progress in understanding the fly's biology and proposing alternative control methods to pesticide use. RESULTS: Genomic DNA was sequenced from male and female Demokritos strain flies, maintained in the laboratory for over 45 years. We used short-, mate-pair-, and long-read sequencing technologies to generate a combined male-female genome assembly (GenBank accession GCA_001188975.2). Genomic DNA sequencing from male insects using 10x Genomics linked-reads technology followed by mate-pair and long-read scaffolding and gap-closing generated a highly contiguous 489 Mb genome with a scaffold N50 of 4.69 Mb and L50 of 30 scaffolds (GenBank accession GCA_001188975.4). RNA-seq data generated from 12 tissues and/or developmental stages allowed for genome annotation. Short reads from both males and females and the chromosome quotient method enabled identification of Y-chromosome scaffolds which were extensively validated by PCR. CONCLUSIONS: The high-quality genome generated represents a critical tool in olive fruit fly research. We provide an extensive RNA-seq data set, and genome annotation, critical towards gaining an insight into the biology of the olive fruit fly. In addition, elucidation of Y-chromosome sequences will advance our understanding of the Y-chromosome's organization, function and evolution and is poised to provide avenues for sterile insect technique approaches.


Asunto(s)
Tephritidae/genética , Cromosoma Y/genética , Cromosoma Y/metabolismo , Animales , Femenino , Genoma de los Insectos/genética , Masculino , Reacción en Cadena de la Polimerasa
11.
Epigenetics Chromatin ; 12(1): 53, 2019 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-31493790

RESUMEN

BACKGROUND: TH2B is a major histone variant that replaces about 80-85% of somatic H2B in mammalian spermatocytes and spermatids. The post-translational modifications (PTMs) on TH2B have been well characterised in spermatocytes and spermatids. However, the biological function(s) of these PTMs on TH2B have not been deciphered in great detail. In our attempt to decipher the unique function(s) of histone variant TH2B, we detected the modification in the N-terminal tail, Serine 11 phosphorylation on TH2B (TH2BS11ph) in spermatocytes. RESULTS: The current study is aimed at understanding the function of the TH2BS11ph modification in the context of processes that occur during meiotic prophase I. Immunofluorescence studies with the highly specific antibodies revealed that TH2BS11ph histone mark is enriched in the unsynapsed axes of the sex body and is associated with XY body-associated proteins like Scp3, γH2AX, pATM, ATR, etc. Genome-wide occupancy studies as determined by ChIP sequencing experiments in P20 C57BL6 mouse testicular cells revealed that TH2BS11ph is enriched in X and Y chromosomes confirming the immunofluorescence staining pattern in the pachytene spermatocytes. Apart from the localisation of this modification in the XY body, TH2BS11ph is majorly associated with H3K4me3-containing genomic regions like gene promoters, etc. These data were also found to corroborate with the ChIP sequencing data of TH2BS11ph histone mark carried out in P12 C57BL6 mouse testicular cells, wherein we found the predominant localisation of this modification at H3K4me3-containing genomic regions. Mass spectrometry analysis of proteins that associate with TH2BS11ph-containing mononucleosomes revealed key proteins linked with the functions of XY body, pericentric heterochromatin and transcription. CONCLUSIONS: TH2BS11ph modification is densely localised in the unsynapsed axes of the XY body of the pachytene spermatocyte. By ChIP sequencing studies in mouse P12 and P20 testicular cells, we demonstrate that TH2BS11ph is predominantly associated with H3K4me3 positive genomic regions like gene promoters, etc. We propose that TH2BS11ph modification could act alone or in concert with other histone modifications to recruit the appropriate transcription or XY body recombination protein machinery at specific genomic loci.


Asunto(s)
Histonas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Estudio de Asociación del Genoma Completo , Código de Histonas , Masculino , Ratones , Ratones Endogámicos C57BL , Fase Paquiteno , Fosforilación , Ratas , Ratas Wistar , Espermatocitos/citología , Espermatocitos/metabolismo , Testículo/citología , Testículo/metabolismo , Cromosoma X/genética , Cromosoma X/metabolismo , Cromosoma Y/genética , Cromosoma Y/metabolismo
12.
Mol Genet Metab ; 127(1): 31-44, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31097364

RESUMEN

Histone demethylases remove transcriptional repressive marks from histones in the nucleus. KDM6A (also known as UTX) is a lysine demethylase which acts on the trimethylated lysine at position 27 in histone 3. The KDM6A gene is located on the X chromosome but escapes X inactivation even though it is not located in the pseudoautosomal region. There is a homologue of KDM6A on the Y chromosome, known as UTY. UTY was thought to have lost its demethylase activity and to represent a non-functional remnant of the ancestral KDM6A gene. However, results with knockout mice suggest that the gene is expressed and the protein performs some function within the cell. Female mice with homozygous deletion of Kdm6a do not survive, but hemizygous males are viable, attributed to the presence of the Uty gene. KDM6A is mutated in the human condition Kabuki syndrome type 2 (OMIM 300867) and in many cases of cancer. The amino acid sequence of KDM6A has been conserved across animal phyla, although it is only found on the X chromosome in eutherian mammals. In this review, we reanalyse existing data from various sources (protein sequence comparison, evolutionary genetics, transcription factor binding and gene expression analysis) to determine the function, expression and evolution of KDM6A and UTY and show that UTY has a functional role similar to KDM6A in metabolism and development.


Asunto(s)
Histona Demetilasas/genética , Histonas/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Proteínas Nucleares/genética , Secuencia de Aminoácidos , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas/metabolismo , Histonas/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Inactivación del Cromosoma X/genética , Cromosoma Y/genética , Cromosoma Y/metabolismo
13.
Heredity (Edinb) ; 123(3): 419-428, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31028370

RESUMEN

Deleterious mutations accumulating on non-recombining Y chromosomes can drive XY to XY turnovers, as they allow to replace the old mutation-loaded Y by a new mutation-free one. The same process is thought to prevent XY to ZW turnovers, because the latter requires fixation of the ancestral Y, assuming dominance of the emergent feminizing mutation. Using individual-based simulations, we explored whether and how an epistatically dominant W allele can spread in a young XY system that gradually accumulates deleterious mutations. We also investigated how sexually antagonistic (SA) polymorphism on the ancestral sex chromosomes and the mechanism controlling X-Y recombination suppression affect these transitions. In contrast with XY to XY turnovers, XY to ZW turnovers cannot be favored by Y chromosome mutation load. If the arrest of X-Y recombination depends on genotypic sex, transitions are strongly hindered by deleterious mutations, and totally suppressed by very small SA cost, because deleterious mutations and female-detrimental SA alleles would have to fix with the Y. If, however, the arrest of X-Y recombination depends on phenotypic sex, X and Y recombine in XY ZW females, allowing for the purge of Y-linked deleterious mutations and loss of the SA polymorphism, causing XY to ZW turnovers to occur at the same rate as in the absence of deleterious and sex-antagonistic mutations. We generalize our results to other types of turnovers (e.g., triggered by non-dominant sex-determining mutations) and discuss their empirical relevance.


Asunto(s)
Anuros/genética , Drosophila melanogaster/genética , Recombinación Genética , Procesos de Determinación del Sexo , Cromosoma X/metabolismo , Cromosoma Y/metabolismo , Alelos , Animales , Epistasis Genética , Femenino , Células Germinativas , Masculino , Modelos Genéticos , Mutación , Selección Genética
14.
Biol Reprod ; 100(3): 810-823, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30285093

RESUMEN

We previously demonstrated that in the mouse only two Y chromosome genes are required for a male to produce an offspring with the help of assisted reproduction technologies (ART): testis determinant Sry and spermatogonial proliferation factor Eif2s3y. Subsequently, we have shown that the function of these genes can be replaced by transgenic overexpression of their homologs, autosomally encoded Sox9 and X-chromosome encoded Eif2s3x. Males with Y chromosome contribution limited to two (XEif2s3yOSry), one (XEif2s3yOSox9 and XOSry,Eif2s3x), and no genes (XOSox9,Eif2s3x) produced haploid germ cells and sired offspring after ART. However, despite successful assisted reproductive outcome, they had smaller testes and displayed abnormal development of the seminiferous epithelium and testicular interstitium. Here we explored whether these testicular defects originated from altered pro-testis and pro-ovary factor signaling in genital ridges at the time of sex determination. Timed pregnancies were generated to obtain transgenic XEif2s3yOSry, XEif2s3yOSox9, XOSry,Eif2s3x, XOSox9,Eif2s3x, and wild-type XX and XY fetuses at 12.5 days post coitum. Dissected genital ridges were assessed for their morphology and anatomy, and expression of pro-testis and pro-ovary transcripts. All transgenic males displayed incomplete masculinization of gonadal shape, impaired development of testicular cords and gonadal vasculature, and decreased expression of factors promoting male pathway. Fetal gonad masculinization was more effective when sex determination was driven by the Sry transgene, in the presence of Y chromosome genes, and to a lesser extent a double dosage of X genes. The study adds to the understanding of the role of Y chromosome genes and their homologs during sex determination.


Asunto(s)
Desarrollo Embrionario/genética , Procesos de Determinación del Sexo/genética , Cromosoma Y/metabolismo , Animales , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica , Genotipo , Masculino , Ratones , Ratones Transgénicos , ARN , Procesos de Determinación del Sexo/fisiología
15.
Anim Reprod Sci ; 198: 121-128, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30274742

RESUMEN

The identification of distinct proteins present on the membrane of spermatozoa with X and Y chromosomes allows the development of immuno-sexing techniques. The aim of this study, therefore, was to use mass spectrometry to analyze the protein profile of sperm previously categorized using flow cytometry into X or Y-bearing semen pools. Sex-sorted sperm samples (n = 6 X and n = 6 Y) were used. Proteins were extracted and analyzed by mass spectrometry using data independent acquisition (DIA). The data were searched against taxonomy Bos taurus in the Swiss Prot database. In total, 459 protein groups were identified. Of these, eight proteins were in differential abundances between the X- and Y-bearing sperm population. Among the major proteinsdetected, EF-hand domain-containing protein 1, a protein involved in embryonic development, is more abundant in Y-bearing spermatozoa. In addition, proteins FUN14, domain-containing protein 2, NADH dehydrogenase [ubiquinone] iron-sulfur protein 7 mitochondrial, cytochrome C oxidase subunit 2, acetyl -CoA carboxylase type beta were more abundant in X-bearing sperm. In conclusion, there were differences in abundance of proteins between X- and Y-bearing bull spermatozoa. This fact, may contribute to future studies related to sperm physiology and possibility development of immuno-sexing techniques.


Asunto(s)
Espectrometría de Masas/métodos , Proteoma/análisis , Preselección del Sexo , Espermatozoides/citología , Espermatozoides/metabolismo , Cromosoma X/metabolismo , Cromosoma Y/metabolismo , Animales , Bovinos , Separación Celular/métodos , Citometría de Flujo/métodos , Inmunoensayo , Masculino , Proteómica , Preselección del Sexo/veterinaria , Espermatozoides/química
16.
Stem Cell Res Ther ; 9(1): 38, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29444715

RESUMEN

BACKGROUND: Wharton's jelly cells (WJCs) have multiple differentiation potentials and are easily harvested in large numbers. WJCs are well tolerated in allogeneic environments and there is a growing list of their therapeutic effects. Most therapies require administering large numbers of cells and this is generally accomplished by intravenous injection. Here, we studied the locations of porcine WJCs in immune-competent, allogeneic hosts after intraperitoneal (IP) injection. METHODS: Male porcine WJCs were administered to female neonatal piglets by IP injection. The location of transplanted cells was examined at 6 h, 24 h, and 7 days after administration using confocal microscopy and polymerase chain reaction (PCR). Transplanted cells were also retrieved from the intestines of recipients and were cultured. Previously transplanted cells were identified by fluorescence in-situ hybridization (FISH) using a Y-chromosome probe. RESULTS: Allogeneic cells were identified in the small and large intestine, stomach, liver, spleen, diaphragm, omentum, kidney, pancreas, mesenteric lymph nodes, heart, lungs, uterus, bladder, and skeletal muscle. Male cells (SRY positive) were found in cultures of cells harvested from the intestinal mucosa 1 week after administration of male porcine WJCs. CONCLUSIONS: Our results show that porcine WJCs distribute widely to the organs in immunocompetent allogeneic hosts after IP administration. They may distribute through the lymphatics initially, and a prominent site of incorporation is the mucosa of the gastrointestinal tract. In that location they could function in the niche of endogenous stem cells and provide secretory products to cells in the tissue damaged by intestinal disease.


Asunto(s)
Rastreo Celular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Aloinjertos , Animales , Femenino , Hibridación Fluorescente in Situ , Masculino , Células Madre Mesenquimatosas/metabolismo , Microscopía Confocal , Reacción en Cadena de la Polimerasa , Porcinos , Cromosoma Y/metabolismo
17.
FASEB J ; 32(5): 2747-2756, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29301944

RESUMEN

The essential role of the Y chromosome in male sex determination has largely overshadowed the possibility that it may exert other biologic roles. Here, we show that Y-chromosome lineage is a strong determinant of perivascular and renal T-cell infiltration in the stroke-prone spontaneously hypertensive rat, which, in turn, may influence vascular function and blood pressure (BP). We also show, for the first time to our knowledge, that augmented perivascular T-cell levels can directly instigate vascular dysfunction, and that the production of reactive oxygen species that stimulate cyclo-oxygenase underlies this. We thus provide strong evidence for the consideration of Y-chromosome lineage in the diagnosis and treatment of male hypertension, and point to the modulation of cardiovascular organ T-cell infiltration as a possible mechanism that underpins Y- chromosome regulation of BP.-Khan, S. I., Andrews, K. L., Jackson, K. L., Memon, B., Jefferis, A.-M., Lee, M. K. S., Diep, H., Wei, Z., Drummond, G. R., Head, G. A., Jennings, G. L., Murphy, A. J., Vinh, A., Sampson, A. K., Chin-Dusting, J. P. F. Y-chromosome lineage determines cardiovascular organ T-cell infiltration in the stroke-prone spontaneously hypertensive rat.


Asunto(s)
Presión Sanguínea , Hipertensión/metabolismo , Hipertensión/fisiopatología , Linfocitos T/metabolismo , Cromosoma Y/metabolismo , Animales , Hipertensión/genética , Masculino , Ratas , Ratas Endogámicas SHR , Ratas Transgénicas , Linfocitos T/patología , Cromosoma Y/genética
18.
Chromosoma ; 127(2): 269-278, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29372309

RESUMEN

Heteromorphic sex chromosomes are common in eukaryotes and largely ubiquitous in birds and mammals. The largest number of multiple sex chromosomes in vertebrates known today is found in the monotreme platypus (Ornithorhynchus anatinus, 2n = 52) which exhibits precisely 10 sex chromosomes. Interestingly, fish, amphibians, and reptiles have sex determination mechanisms that do or do not involve morphologically differentiated sex chromosomes. Relatively few amphibian species carry heteromorphic sex chromosomes, and when present, they are frequently represented by only one pair, either XX:XY or ZZ:ZW types. Here, in contrast, with several evidences, from classical and molecular cytogenetic analyses, we found 12 sex chromosomes in a Brazilian population of the smoky jungle frog, designated as Leptodactylus pentadactylus Laurenti, 1768 (Leptodactylinae), which has a karyotype with 2n = 22 chromosomes. Males exhibited an astonishing stable ring-shaped meiotic chain composed of six X and six Y chromosomes. The number of sex chromosomes is larger than the number of autosomes found, and these data represent the largest number of multiple sex chromosomes ever found among vertebrate species. Additionally, sequence and karyotype variation data suggest that this species may represent a complex of species, in which the chromosomal rearrangements may possibly have played an important role in the evolution process.


Asunto(s)
Anuros/genética , Procesos de Determinación del Sexo , Cromosoma X/metabolismo , Cromosoma Y/metabolismo , Animales , Anuros/clasificación , Brasil , Hibridación Genómica Comparativa , Femenino , Hibridación Fluorescente in Situ , Cariotipo , Cariotipificación , Masculino , Filogenia , Cromosoma X/ultraestructura , Cromosoma Y/ultraestructura
19.
Chromosoma ; 127(2): 261-267, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29256059

RESUMEN

X inactivation is a fundamental mechanism in eutherian mammals to restore a balance of X-linked gene products between XY males and XX females. However, it has never been extensively studied in a eutherian species with a sex determination system that deviates from the ubiquitous XX/XY. In this study, we explore the X inactivation process in the African pygmy mouse Mus minutoides, that harbours a polygenic sex determination with three sex chromosomes: Y, X, and a feminizing mutant X, named X*; females can thus be XX, XX*, or X*Y, and all males are XY. Using immunofluorescence, we investigated histone modification patterns between the two X chromosome types. We found that the X and X* chromosomes are randomly inactivated in XX* females, while no histone modifications were detected in X*Y females. Furthermore, in M. minutoides, X and X* chromosomes are fused to different autosomes, and we were able to show that the X inactivation never spreads into the autosomal segments. Evaluation of X inactivation by immunofluorescence is an excellent quantitative procedure, but it is only applicable when there is a structural difference between the two chromosomes that allows them to be distinguished.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Procesamiento Proteico-Postraduccional , Procesos de Determinación del Sexo , Inactivación del Cromosoma X , Cromosoma X/metabolismo , Cromosoma Y/metabolismo , Acetilación , África , Animales , Animales Salvajes , Cromosomas de los Mamíferos/ultraestructura , Femenino , Técnica del Anticuerpo Fluorescente/métodos , Histonas/genética , Histonas/metabolismo , Cariotipificación , Masculino , Metilación , Ratones , Cromosoma X/ultraestructura , Cromosoma Y/ultraestructura
20.
J Cell Physiol ; 233(9): 6591-6603, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29115663

RESUMEN

Annexin A1 (AnxA1) is a glucocorticoid-regulated anti-inflammatory protein secreted by phagocytes and other specialised cells. In the endocrine system, AnxA1 controls secretion of steroid hormones and it is abundantly expressed in the testis, ovaries, placenta and seminal fluid, yet its potential modulation of fertility has not been described. Here, we observed that AnxA1 knockout (KO) mice delivered a higher number of pups, with a higher percentage of female offsprings. This profile was not dependent on the male features, as sperm from KO male mice did not present functional alterations, and had an equal proportion of Y and X chromosomes, comparable to wild type (WT) male mice. Furthermore, mismatched matings of male WT mice with female KO yielded a higher percentage of female pups per litter, a phenomenon which was not observed when male KO mice mated with female WT animals. Indeed, AnxA1 KO female mice displayed several differences in parameters related to gestation including (i) an arrested estrous cycle at proestrus phase; (ii) increased sites of implantation; (iii) reduced pre- and post-implantation losses; (iv) exacerbated features of the inflammatory reaction in the uterine fluid during implantation phase; and (v) enhanced plasma progesterone in the beginning of pregnancy. In summary, herein we highlight that AnxA1 pathway as a novel determinant of fundamental non-redundant regulatory functions during early pregnancy.


Asunto(s)
Anexina A1/metabolismo , Implantación del Embrión/fisiología , Animales , Ciclo Estral/metabolismo , Ciclo Estral/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Modelos Animales , Embarazo , Proestro/metabolismo , Proestro/fisiología , Razón de Masculinidad , Útero/metabolismo , Útero/fisiología , Cromosoma X/metabolismo , Cromosoma X/fisiología , Cromosoma Y/metabolismo , Cromosoma Y/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...